Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Am J Hematol ; 87(2): 172-4, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22081487

RESUMEN

Patients undergoing allogeneic hematopoietic stem cell transplant (allo HCT) have a higher incidence of infections partly due to secondary hypogammaglobulinemia. We evaluated the role of IVIG in allo HCT patients who received prophylactic IVIG 200 mg/kg once weekly regardless of IgG level (Group 1, n = 115) compared with patients who received IVIG based on IgG level <400 mg/dL (Group 2, n = 114). Primary endpoints were the utilization of IVIG, incidence of veno-occlusive disease (VOD), graft-versus-host disease (GVHD), and documented infections within the first 100 days after allo HCT. Patients in both groups were similar except for a higher number of matched unrelated donor (MUD) transplants in Group 2 (62 vs. 41, P = 0.01). There were no significant differences in the incidence all grades of GVHD (55 vs. 50), VOD (2 vs. 0) or infections in the two groups except for a higher incidence of para-influenza infections in group 1 (9 vs. 0, P = 0.003) coinciding with the flu season. We recommend monthly monitoring of IgG level and replacement only if IgG level is <400 mg/dL.


Asunto(s)
Agammaglobulinemia/prevención & control , Monitoreo de Drogas , Trasplante de Células Madre Hematopoyéticas , Inmunoglobulinas Intravenosas/uso terapéutico , Infecciones por Respirovirus/prevención & control , Respirovirus/crecimiento & desarrollo , Adolescente , Adulto , Agammaglobulinemia/sangre , Agammaglobulinemia/inmunología , Anciano , Niño , Preescolar , Esquema de Medicación , Medicina Basada en la Evidencia , Femenino , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunoglobulinas Intravenosas/sangre , Inmunoglobulinas Intravenosas/farmacología , Masculino , Persona de Mediana Edad , Infecciones por Respirovirus/sangre , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Estudios Retrospectivos , Trasplante Homólogo
2.
Curr Drug Targets Infect Disord ; 5(4): 401-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16535861

RESUMEN

The enzyme neuraminidase (NA) is an attractive target for antiviral strategy because of its essential role in the pathogenicity of many respiratory viruses. NA removes sialic acid from the surface of infected cells and virus particles, thereby preventing viral self-aggregation and promoting efficient viral spread; NA also plays a role in the initial penetration of the mucosal lining of the respiratory tract. Random screening for inhibitors has identified only low-affinity and nonselective viral NA inhibitors. Selective, high-affinity inhibitors of influenza virus neuraminidase, zanamivir and oseltamivir, were developed using computer-aided design techniques on the basis of the three-dimensional structure of the influenza virus NA. These drugs were highly efficient in inhibiting replication of both influenza A and B viruses in vitro and in vivo and were approved for human use in 1999. Subsequently, the same structure-based design approach was used for the rational design of inhibitors of the parainfluenza virus hemagglutinin-neuraminidase (HN). One of these compounds, BCX 2798, effectively inhibited NA activity, cell binding, and growth of parainfluenza viruses in tissue culture and in the lungs of infected mice. Clinical reports indicate high efficiency of NA inhibitors for prophylaxis and treatment of influenza virus infection, good tolerance, and a low rate of emergence of drug-resistant mutants. Future experimental and clinical studies should establish the viability of NA inhibitors for the treatment of other respiratory virus infections.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Diseño de Fármacos , Neuraminidasa/antagonistas & inhibidores , Acetamidas/química , Acetamidas/farmacología , Animales , Azidas/química , Azidas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Guanidinas/química , Guanidinas/farmacología , Proteína HN/química , Proteína HN/inmunología , Ácidos Hexurónicos/química , Ácidos Hexurónicos/farmacología , Humanos , Estructura Molecular , Neuraminidasa/química , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/enzimología , Orthomyxoviridae/crecimiento & desarrollo , Oseltamivir , Conformación Proteica , Piranos/química , Piranos/farmacología , Respirovirus/efectos de los fármacos , Respirovirus/enzimología , Respirovirus/crecimiento & desarrollo , Ácidos Siálicos/química , Ácidos Siálicos/farmacología , Zanamivir
3.
Clin Diagn Lab Immunol ; 11(3): 599-607, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15138188

RESUMEN

Defensins and surfactant protein A (SP-A) and SP-D are antimicrobial components of the pulmonary innate immune system. The purpose of this study was to determine the extent to which parainfluenza type 3 virus infection in neonatal lambs alters expression of sheep beta-defensin 1 (SBD-1), SP-A, and SP-D, all of which are constitutively transcribed by respiratory epithelia. Parainfluenza type 3 viral antigen was detected by immunohistochemistry (IHC) in the bronchioles of all infected lambs 3 days postinoculation and at diminished levels 6 days postinoculation, but it was absent 17 days postinoculation. At all times postinoculation, lung homogenates from parainfluenza type 3 virus-inoculated animals had increased SBD-1, SP-A, and SP-D mRNA levels as detected by fluorogenic real-time reverse transcriptase PCR. Protein levels of SP-A in lung homogenates detected by quantitative-competitive enzyme-linked immunosorbent assay and protein antigen of SP-A detected by IHC were not altered. These studies demonstrate that parainfluenza type 3 virus infection results in enhanced expression of constitutively transcribed innate immune factors expressed by respiratory epithelia and that this increased expression occurs concurrently with decreased viral replication.


Asunto(s)
Infecciones por Paramyxoviridae/inmunología , Neumonía/inmunología , Proteína A Asociada a Surfactante Pulmonar/genética , ARN Mensajero/metabolismo , Replicación Viral , beta-Defensinas/genética , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/sangre , Antígenos Virales/análisis , Antígenos Virales/inmunología , Temperatura Corporal , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/química , Células Epiteliales/virología , Femenino , Expresión Génica , Inmunohistoquímica , Pulmón/química , Pulmón/patología , Pulmón/virología , Macrófagos Alveolares/química , Macrófagos Alveolares/virología , Masculino , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/veterinaria , Neumonía/genética , Neumonía/veterinaria , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/genética , ARN Mensajero/genética , Respirovirus/química , Respirovirus/crecimiento & desarrollo , Respirovirus/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ovinos , Enfermedades de las Ovejas/genética , Enfermedades de las Ovejas/inmunología , Enfermedades de las Ovejas/virología
4.
Antiviral Res ; 60(3): 209-19, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14638397

RESUMEN

A cell-based assay was used to discover compounds inhibiting respiratory syncytial virus (RSV)-induced fusion in HeLa/M cells. A lead compound was identified and subsequent synthesis of >300 analogues led to the identification of JNJ 2408068 (R170591), a low molecular weight (MW 395) benzimidazole derivative with an EC(50) (0.16 nM) against some lab strains almost 100,000 times better than that of ribavirin (15 microM). Antiviral activity was confirmed for subgroup A and B clinical isolates of human RSV and for a bovine RSV isolate. The compound did not inhibit the growth of representative viruses from other Paramyxovirus genera, i.e. HPIV2 and Mumps Virus (genus Rubulavirus), HPIV3 (genus Respirovirus), Measles virus (genus Morbillivirus) and hMPV. Efficacy in cytopathic effect inhibition assays correlated well with efficacy in virus yield reduction assays. A concentration of 10nM reduced RSV production 1000-fold in multi-cycle experiments, irrespective of the multiplicity of infection. Time of addition studies pointed to a dual mode of action: inhibition of virus-cell fusion early in the infection cycle and inhibition of cell-cell fusion at the end of the replication cycle. Two resistant mutants were raised and shown to have single point mutations in the F-gene (S398L and D486N). JNJ 2408068 was also shown to inhibit the release of proinflammatory cytokines IL-6, IL-8 and Rantes from RSV-infected A549 cells.


Asunto(s)
Antivirales/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Antivirales/química , Fusión Celular , Citocinas/metabolismo , Efecto Citopatogénico Viral/efectos de los fármacos , Análisis Mutacional de ADN , Farmacorresistencia Viral/genética , Células HeLa , Humanos , Metapneumovirus/efectos de los fármacos , Metapneumovirus/crecimiento & desarrollo , Peso Molecular , Morbillivirus/efectos de los fármacos , Morbillivirus/crecimiento & desarrollo , Mutación Puntual , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Virus Sincitiales Respiratorios/aislamiento & purificación , Virus Sincitiales Respiratorios/patogenicidad , Respirovirus/efectos de los fármacos , Respirovirus/crecimiento & desarrollo , Rubulavirus/efectos de los fármacos , Rubulavirus/crecimiento & desarrollo , Proteínas Virales de Fusión/genética , Ensayo de Placa Viral
5.
J Appl Microbiol ; 91(4): 750-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11576313

RESUMEN

AIMS: This study was carried out to determine the survival time of Escherichia coli, Salmonella choleraesuis, Aujeszky's Disease virus and Blue Eye Disease virus in ensilages based on the solid fraction of pig faeces. METHODS AND RESULTS: The four micro-organisms were inoculated into microsilos based on the solid fraction of pig faeces, sorghum and molasses. They were left for 0, 7, 14, 28 and 56 days, after which the state of each microsilo was evaluated, and isolation of the inoculated agents was attempted. The four inoculated agents were isolated only on day 0 of ensilage. The viral agents were identified through the cytopathic effect and fluorescence. CONCLUSIONS, SIGNIFICANCE AND IMPACT OF THE STUDY: It is concluded that ensilages based on the solid fraction of pig faeces appear to reduce the risk of the transmission of the agents inoculated in this study and help to reduce the environmental impact by using the solid in animal feed.


Asunto(s)
Escherichia coli/crecimiento & desarrollo , Herpesvirus Suido 1/crecimiento & desarrollo , Respirovirus/crecimiento & desarrollo , Salmonella/crecimiento & desarrollo , Ensilaje/microbiología , Ensilaje/virología , Animales , Escherichia coli/clasificación , Heces/microbiología , Heces/virología , Salmonella/clasificación , Porcinos
6.
Arch Virol ; 146(5): 893-908, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11448028

RESUMEN

We investigated the mechanisms responsible for attenuation of mouse pathogenicity of Sendai virus (SeV) through passages in eggs. A highly virulent clone, E0, derived from the field SeV Hamamatsu strain, was successively passaged in hen's eggs. Analysis of the mouse lethal dose 50% (MLD50) of virus clones obtained from the viruses at egg-passages 1, 15, 30 and 50 demonstrated that attenuation of E0 by egg-passage occurred due to the gradual appearance of and replacement by virus variants possessing higher MLD50. Comparison of viral replication in the mouse lung and mouse pathogenicity with the representative SeV clones, E0, E15c12, E30c12 and E50c19, obtained from the respective egg-passages revealed that the low pathogenicity of the egg-passaged clones was due to poor multi-cycle viral replication in the lung. Furthermore, MLD50s of the SeV clones were found to be negatively correlated with the replication capability in primary mouse pulmonary epithelial (MPE) cells; the egg-passaged clones with more attenuated phenotypes showed lower replication capability in MPE cells. In the MPE cells infected with the SeV clones at m.o.i. 10, however, viral protein and mRNA syntheses of the egg-passaged clones were enhanced or comparable to those of the parental E0 clone at 1 day and 2 days post infection (p.i.) but decreased more rapidly thereafter. In contrast, viral genome synthesis of the egg-passaged clones in the cells at 2 days p.i. was several times lower than that of E0. These results strongly suggest that attenuation of a virulent field SeV strain by egg-passage occurs due to the appearance and selection of virus variants possessing poor propagation capacity in mouse respiratory epithelial cells, which is caused primarily by an impediment of viral genome replication.


Asunto(s)
Pulmón/virología , Infecciones por Respirovirus/virología , Respirovirus/patogenicidad , Replicación Viral , Animales , Línea Celular , Embrión de Pollo , Genoma Viral , Riñón , Dosificación Letal Mediana , Pulmón/patología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos ICR , Fenotipo , ARN Viral/biosíntesis , Respirovirus/genética , Respirovirus/crecimiento & desarrollo , Especificidad de la Especie , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Virulencia , Cultivo de Virus , Pérdida de Peso
7.
Virus Genes ; 20(2): 173-82, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10872880

RESUMEN

The Kansas/15626/84 (Ka) and Shipping Fever (SF) strains of bovine parainfluenza virus type 3 (BPIV3) replicate less efficiently than human PIV3 (HPIV3) in the upper and lower respiratory tract of rhesus monkeys, and BPIV3 Ka is also highly attenuated in humans and is in clinical trials as a candidate vaccine against HPIV3. To initiate an investigation of the genetic basis of the observed attenuation phenotype of BPIV3 in primates, the complete genomic sequences of Ka and SF genomes were determined and compared to those of BPIV3 strain 910N and two HPIV3 strains, JS and Wash/47885/57. There is a high degree of identity between the five PIV3 viruses in their 55 nucleotide (nt) leader (83.6%) and 44 nt trailer (93.2%) sequences. The five viruses display amino acid sequence identity ranging from 58.6% for the phosphoprotein to 89.7% for the matrix protein. Interestingly, the majority of amino acid residues found to be variable at a given position in a five-way protein alignment are nonetheless identical within the viruses of either host species (BPIV3 or HPIV3). These host-specific residues might be products of distinct selective pressures on BPIV3 and HPIV3 during evolution in their respective hosts. These host-specific sequences likely include ones which are responsible for the host range differences, such as the efficient growth of BPIV3 in bovines compared to its restricted growth in primates. It should now be possible using the techniques of reverse genetics to import sequences from BPIV3 into HPIV3 and identify those nt or protein sequences which attenuate HPIV3 for primates. This information should be useful in understanding virus-host interactions and in the development of vaccines to protect against HPIV3-induced disease.


Asunto(s)
Genoma Viral , Macaca mulatta/virología , Respirovirus/genética , Replicación Viral , Animales , Secuencia de Bases , Bovinos , Línea Celular , Secuencia de Consenso , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 3 Humana/crecimiento & desarrollo , Virus de la Parainfluenza 3 Humana/fisiología , ARN Viral/análisis , Respirovirus/crecimiento & desarrollo , Respirovirus/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
8.
Artículo en Inglés | MEDLINE | ID: mdl-10763377

RESUMEN

The reptilian paramyxovirus GOV was successfully adapted to Vero cells in 80 passages at 30 degrees C. The virus replicated with HA titres of 1:64 and 10(7.5) TCID50/ml in the heterologous host cells forming syncytia, giant cells and cytoplasmic inclusion bodies. After 80 passages GOV was identified by immunofluorescence, by immune electron microscopy and by PCR. Sequencing of RT-PCR products of GOV after 80 passages did not reveal changes in the nucleotide sequence of the partial L-gene of GOV.


Asunto(s)
Colubridae/virología , Infecciones por Paramyxoviridae/veterinaria , Respirovirus/crecimiento & desarrollo , Animales , Chlorocebus aethiops , Efecto Citopatogénico Viral , ADN Viral/química , Técnica del Anticuerpo Fluorescente Indirecta/veterinaria , Pruebas de Hemaglutinación/veterinaria , Microscopía Inmunoelectrónica/veterinaria , Infecciones por Paramyxoviridae/virología , Respirovirus/genética , Respirovirus/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Análisis de Secuencia de ADN , Células Vero
9.
Virology ; 267(2): 209-19, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10662616

RESUMEN

P56 is the most abundant protein induced by interferon (IFN) treatment of human cells. To facilitate studies on its induction pattern and cellular functions, we expressed recombinant P56 as a hexahistidine-tagged protein in Escherichia coli and purified it to apparent homogeneity using affinity chromatography. A polyclonal antibody raised against this recombinant protein was used to show that P56 is primarily a cytoplasmic protein. Cellular expression of P56 by transfection did not inhibit the replication of vesicular stomatitis virus and encephalomyocarditis virus. P56 synthesis was rapidly induced by IFN-beta, and the protein had a half-life of 6 h. IFN-gamma or poly(A)(+) could not induce the protein, but poly(I)-poly(C) or an 85-bp synthetic double-stranded RNA efficiently induced it. Similarly, infection of GRE cells, which are devoid of type I IFN genes, by vesicular stomatitis virus, encephalomyocarditis virus, or Sendai virus caused P56 induction. Surprisingly, Sendai virus could also induce P56 in the mutant cell line P2.1, which cannot respond to either IFN-alpha/beta or double-stranded RNA. Induction of P56 in the P2.1 cells and the parental U4C cells by virus infection was preceded by activation of IRF-3 as judged by its translocation to the nucleus from the cytoplasm.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Interferones/farmacología , ARN Bicatenario/farmacología , Anticuerpos Monoclonales/inmunología , Línea Celular , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Humanos , Interferón beta/farmacología , Proteínas Recombinantes/genética , Respirovirus/crecimiento & desarrollo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/virología , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo
10.
Proc Natl Acad Sci U S A ; 96(21): 11860-5, 1999 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-10518541

RESUMEN

Persistent infections by viruses such as HIV-1 and hepatitis B virus can pose long-term health hazards. Because establishment of persistent infections involves close interactions and adjustments in both host and virus, it would be informative to establish a paradigm with which a normally cytolytic viral infection can be easily converted to persistent infection, so that the different stages in developing persistent infection can be examined. Such a model system is described in this paper. Highly cytolytic encephalomyocarditis virus (EMCV) infection was shifted to persistent infection as a result of repressed expression of the double-stranded RNA-dependent protein kinase (PKR) in the promonocytic U937 cells. Because of the apoptogenic potential of PKR, a deficiency of PKR resulted in a delay in virus-induced apoptosis in EMCV-infected U937 cells, allowing the eventual establishment of persistent EMCV infection in these cells (U9K-AV2). That this was a bona fide persistent infection was demonstrated by the ability of infected cells to propagate as long-term virus-shedding cultures; electron microscopy studies showing presence of intracellular EMCV virions and chromatin condensation; detection of virus-induced chromosomal DNA fragmentation and sustained expression of apoptogenic p53 and IL-1beta converting enzyme; and demonstration of active EMCV transcription by reverse transcription-PCR. In addition, a host-virus coevolution was observed in U9K-AV2 cultures over time: U9K-AV2 cells exhibited slower growth rates, resistance to viral super-infection, and cessation of IFN-alpha synthesis, whereas the infectivity of EMCV was drastically attenuated. Finally, data are presented on the suitability of this model to study establishment of persistent infection by other viruses such as Sendai virus and reovirus.


Asunto(s)
Apoptosis , Virus de la Encefalomiocarditis/crecimiento & desarrollo , eIF-2 Quinasa/fisiología , Supervivencia Celular , Efecto Citopatogénico Viral , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferones/metabolismo , Modelos Biológicos , ARN sin Sentido/metabolismo , Reoviridae/crecimiento & desarrollo , Respirovirus/crecimiento & desarrollo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Células U937 , Activación Viral
12.
J Virol ; 73(8): 6228-34, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10400712

RESUMEN

Simian parainfluenza virus 5 (SV5) is a prototype of the Paramyxoviridae family of nonsegmented negative-sense RNA viruses. The single-stranded RNA genomes of these viruses contain a series of tandemly linked genes separated by intergenic (IG) sequences flanked by gene-end (GE) and gene-start (GS) sequences. The viral RNA polymerase (vRNAP) complex is thought to enter the genome at its 3' end, and synthesis of mRNAs is thought to occur by a stop-start mechanism in a sequential and polar manner, with transcriptional attenuation occurring primarily at the intergenic regions. As a result, multiple nonoverlapping mRNA species are generated for each single entry of the vRNAP. To investigate the functions of GE, IG, and GS sequences in transcription, we constructed plasmids containing cDNAs of the full-length SV5 genome in which the gene junction sequences (GE, IG, and GS sequences) located between the hemagglutinin-neuraminidase (HN) and the polymerase (L) genes were replaced with the counterpart sequences from other gene junctions. By using reverse genetics, we recovered viable viruses from each cDNA construct, although their growth characteristics varied. Analysis of the HN and L mRNAs by quantitative RNase protection assay indicated that the ratios of HN to L mRNAs varied over a fourfold range. The alteration of the gene junction sequences also permitted examination of the hypothesized requirement for hexamer nucleotide position of the GS sites. The recovery of infectious viruses with transcription initiation sites that occurred at nucleotide positions 1, 2, 3, 5, and 6 of the hexamer suggest that the requirement is nonstringent.


Asunto(s)
Genes Virales , Proteína HN/genética , Mutagénesis Insercional , Respirovirus/genética , Proteínas Virales/genética , Animales , Sitios de Unión , Bovinos , Línea Celular , Chlorocebus aethiops , Cricetinae , Expresión Génica , Humanos , Fenotipo , ARN Mensajero , Respirovirus/crecimiento & desarrollo , Transcripción Genética , Células Tumorales Cultivadas
13.
J Gen Virol ; 80 ( Pt 6): 1393-1399, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10374956

RESUMEN

The capacity of DNA vaccines to prime CD8+ T cells makes them excellent candidates for vaccines that are designed to emphasize cellular immunity. However, the long-term stability of CD8+ T cell memory induced by DNA vaccination is poorly characterized. Here, the quality of CD8+ T cell recall responses in mice was investigated more than 1 year after DNA vaccination with the Sendai virus nucleoprotein gene. Cytotoxic T lymphocyte (CTL) activity specific for both dominant and subdominant epitopes could be recalled readily 1 year after vaccination and the frequencies of CTL precursors specific for both of these epitopes were relatively high. These CTL responded strongly to subsequent Sendai virus infection in terms of their ability to migrate to the lung and to differentiate into effector cells. In addition, the recall response to virus infection, as determined by CTL activity in the lungs and IFN-gamma responses in the spleen, was both faster and greater in magnitude than that in control-immunized mice. Significantly, virus titres were reduced at least 100-fold in the lungs of mice that were immunized more than 1 year before infection, as compared with control mice. These data demonstrate that CD8+ T cell memory elicited by DNA vaccination is functionally relevant and persists for at least 1 year.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Nucleoproteínas , Infecciones por Respirovirus/inmunología , Respirovirus/inmunología , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Femenino , Epítopos Inmunodominantes/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Respirovirus/crecimiento & desarrollo , Infecciones por Respirovirus/prevención & control , Infecciones por Respirovirus/virología , Linfocitos T Citotóxicos/inmunología , Vacunación , Vacunas de ADN/administración & dosificación , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/uso terapéutico , Vacunas Virales/administración & dosificación
14.
J Immunol Methods ; 222(1-2): 189-96, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10022385

RESUMEN

The expression of type I interferons (IFNs) in eukaryotic cells represents a first line of defense against viral infection. Cells pretreated by IFNs do not support viral replication and are protected from virus-induced cell destruction. A challenge of IFN-pretreated cells with vesicular stomatitis virus (VSV) is frequently used to quantitate this cytokine because, on the one hand, the replication of VSV is highly sensitive to IFNs and, on the other hand, in unprotected cells this virus induces a rapid cytopathic effect that can readily be quantified. However, as VSV may infect humans and is known to cause severe disease in a variety of animal species, this virus must be considered a biohazard. In this paper, we describe a bioassay for bovine IFN using Sendai virus, a paramyxovirus that grows readily in MDBK cells yet is released from these cells in a non-infectious form. The sensitivity and dynamic range of this assay are similar to those of the popular VSV-based IFN assay. We demonstrate that the Sendai-virus-based IFN assay permits rapid quantitation of recombinant bovine type I IFN, and also of native type I IFNs which are present in the supernatants of monocyte-derived macrophages infected with various pathogens. In view of the possible artifacts induced by viruses in samples to be assayed for IFN activity, we evaluated several methods of virus inactivation. Treatment with beta-propiolactone led to virus inactivation without affecting the bioactivity of IFNs as detected in the Sendai-virus-based assay.


Asunto(s)
Antivirales/farmacología , Interferón Tipo I/farmacología , Respirovirus/efectos de los fármacos , Respirovirus/crecimiento & desarrollo , Animales , Antiinfecciosos Locales/farmacología , Bovinos , Células Cultivadas , Riñón/virología , Propiolactona/farmacología , Conejos , Sensibilidad y Especificidad , Ultrafiltración , Rayos Ultravioleta , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Activación Viral/efectos de los fármacos
15.
J Biol Regul Homeost Agents ; 13(4): 201-6, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10703943

RESUMEN

About 30 Sendai Virus (SV) preparations, examined for their capacity to induce natural human interferon alpha from fresh human leukocytes (Le-IFN-alpha) of healthy donors, were characterized for hemagglutinating (HA) and hemolytic (HemA) activities and for SDS-PAGE proteic pattern. The SV preparations were produced by a single passage or by serial propagations through eggs in different conditions of multiplicity of infection (m.o.i.). The produced SV subpopulations showed variable IFN-inducing capacity, the values of which are distributed over a 6-fold range resembling a Gaussian distribution. No detectable difference was observed comparing the SV preparation obtained by serial propagations with those obtained by a single passage. The variability of the measured HA activity and HemA activity and as well as the SDS-PAGE proteic pattern of the SV preparations did not correlate with the induced amount of IFN per cell. In the same experimental conditions to produce Le-IFN-alpha, u.v.-treated SV preparations were unable to induce interferon depending on the u.v.-treatment. So it can be concluded that the distinct nHu-IFN-alpha-inducing capacity of SV subpopulation could be mainly associated with divergent compositions of the viral RNAs rather than with a different contents of viral proteins, among those detectable in SDS-PAGE and those responsible for HA activity and for HemA activity.


Asunto(s)
Hemaglutinación por Virus/inmunología , Hemólisis/efectos de los fármacos , Interferón-alfa/biosíntesis , Leucocitos/virología , Respirovirus/inmunología , Proteínas Virales/farmacología , Animales , Células Cultivadas , Embrión de Pollo , Electroforesis en Gel de Poliacrilamida , Pruebas de Hemaglutinación , Hemaglutinación por Virus/efectos de los fármacos , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/farmacología , Humanos , Leucocitos/citología , Leucocitos/metabolismo , ARN Viral/inmunología , ARN Viral/efectos de la radiación , Respirovirus/crecimiento & desarrollo , Respirovirus/efectos de la radiación , Pase Seriado , Rayos Ultravioleta , Ensayo de Placa Viral , Proteínas Virales/biosíntesis , Proteínas Virales/química
16.
J Virol ; 72(12): 9747-54, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9811709

RESUMEN

In the assembly of paramyxoviruses, interactions between viral proteins are presumed to be specific. The focus of this study is to elucidate the protein-protein interactions during the final stage of viral assembly that result in the incorporation of the viral envelope proteins into virions. To this end, we examined the specificity of HN incorporation into progeny virions by transiently transfecting HN cDNA genes into Sendai virus (SV)-infected cells. SV HN expressed from cDNA was efficiently incorporated into progeny Sendai virions, whereas Newcastle disease virus (NDV) HN was not. This observation supports the theory of a selective mechanism for HN incorporation. To identify the region on HN responsible for the selective incorporation, we constructed chimeric SV and NDV HN cDNAs and evaluated the incorporation of expressed proteins into progeny virions. Chimera HN that contained the SV cytoplasmic domain fused to the transmembrane and external domains of the NDV HN was incorporated to SV particles, indicating that amino acids in the cytoplasmic domain are responsible for the observed specificity. Additional experiments using the chimeric HNs showed that 14 N-terminal amino acids are sufficient for the specificity. Further analysis identified five consecutive amino acids (residues 10 to 14) that were required for the specific incorporation of HN into SV. These residues are conserved among all strains of SV as well as those of its counterpart, human parainfluenza virus type 1. These results suggest that this region near the N terminus of HN interacts with another viral protein(s) to lead to the specific incorporation of HN into progeny virions.


Asunto(s)
Proteína HN/genética , Proteína HN/metabolismo , Respirovirus/genética , Respirovirus/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Línea Celular , Citoplasma/virología , ADN Complementario/genética , Proteína HN/química , Humanos , Datos de Secuencia Molecular , Morbillivirus/genética , Fosforilación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Respirovirus/crecimiento & desarrollo , Homología de Secuencia de Aminoácido
17.
Virology ; 250(1): 30-40, 1998 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-9770417

RESUMEN

The SH gene of the paramyxovirus SV5 is located between the genes for the glycoproteins, fusion protein (F) and hemagglutinin-neuraminidase (HN), and the SH gene encodes a small 44-residue hydrophobic integral membrane protein (SH). The SH protein is expressed in SV5-infected cells and is oriented in membranes with its N terminus in the cytoplasm. To study the function of the SH protein in the SV5 virus life cycle, the SH gene was deleted from the infectious cDNA clone of the SV5 genome. By using the recently developed reverse genetics system for SV5, it was found that an SH-deleted SV5 (rSV5DeltaSH) could be recovered, indicating the SH protein was not essential for virus viability in tissue culture. Analysis of properties of rSV5DeltaSH indicated that lack of expression of SH protein did not alter the expression level of the other virus proteins, the subcellular localization of F and HN, or fusion competency as measured by lipid mixing assays and a new content mixing assay that did not require the use of vaccinia virus. The growth rate, infectivity, and plaque size of rSV5 and rSV5DeltaSH were found to be very similar. Although SH is shown to be a component of purified virions by immunoblotting, examination of purified rSV5DeltaSH by electron microscopy did not show an altered morphology from SV5. Thus in tissue culture cells the lack of the SV5 SH protein does not confer a recognizable phenotype.


Asunto(s)
Proteínas de la Membrana/fisiología , Respirovirus/crecimiento & desarrollo , Animales , Fusión Celular , Línea Celular , Eliminación de Gen , Genes Virales , Proteína HN/análisis , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , Respirovirus/ultraestructura , Transfección , Proteínas Virales de Fusión/análisis , Ensayo de Placa Viral , Proteínas Estructurales Virales/genética , Virión/ultraestructura
18.
Mol Cell Biol ; 18(5): 2986-96, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9566918

RESUMEN

The interferon regulatory factors (IRF) consist of a growing family of related transcription proteins first identified as regulators of the alpha beta interferon (IFN-alpha/beta) gene promoters, as well as the interferon-stimulated response element (ISRE) of some IFN-stimulated genes. IRF-3 was originally identified as a member of the IRF family based on homology with other IRF family members and on binding to the ISRE of the ISG15 promoter. IRF-3 is expressed constitutively in a variety of tissues, and the relative levels of IRF-3 mRNA do not change in virus-infected or IFN-treated cells. In the present study, we demonstrate that following Sendai virus infection, IRF-3 is posttranslationally modified by protein phosphorylation at multiple serine and threonine residues, which are located in the carboxy terminus of IRF-3. A combination of IRF-3 deletion and point mutations localized the inducible phosphorylation sites to the region -ISNSHPLSLTSDQ- between amino acids 395 and 407; point mutation of residues Ser-396 and Ser-398 eliminated virus-induced phosphorylation of IRF-3 protein, although residues Ser-402, Thr-404, and Ser-405 were also targets. Phosphorylation results in the cytoplasm-to-nucleus translocation of IRF-3, DNA binding, and increased transcriptional activation. Substitution of the Ser-Thr sites with the phosphomimetic Asp generated a constitutively active form of IRF-3 that functioned as a very strong activator of promoters containing PRDI-PRDIII or ISRE regulatory elements. Phosphorylation also appears to represent a signal for virus-mediated degradation, since the virus-induced turnover of IRF-3 was prevented by mutation of the IRF-3 Ser-Thr cluster or by proteasome inhibitors. Interestingly, virus infection resulted in the association of IRF-3 with the CREB binding protein (CBP) coactivator, as detected by coimmunoprecipitation with anti-CBP antibody, an interaction mediated by the C-terminal domains of both proteins. Mutation of residues Ser-396 and Ser-398 in IRF-3 abrogated its binding to CBP. These results are discussed in terms of a model in which virus-inducible, C-terminal phosphorylation of IRF-3 alters protein conformation to permit nuclear translocation, association with transcriptional partners, and primary activation of IFN- and IFN-responsive genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Procesamiento Proteico-Postraduccional , Respirovirus/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Compartimento Celular , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/metabolismo , Citoplasma/metabolismo , Análisis Mutacional de ADN , Histona Acetiltransferasas , Humanos , Factor 3 Regulador del Interferón , Ratones , Datos de Secuencia Molecular , Complejos Multienzimáticos/metabolismo , Coactivador 3 de Receptor Nuclear , Mapeo Peptídico , Fosforilación , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Transactivadores/metabolismo , Activación Transcripcional
19.
J Virol ; 72(4): 2927-34, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9525613

RESUMEN

An avirulent mutant of Sendai virus, Ohita-MVC11 (MVC11), was generated from a highly virulent field strain, Ohita-M1 (M1), through successive passages in LLC-MK2 cell cultures (M. Itoh, Y. Isegawa, H. Hotta, and M. Homma, J. Gen. Virol. 78:3207-3215, 1997). In LLC-MK2 cells, MVC11 induced a high degree of apoptotic cell death that was demonstrated by chromatin condensation of the nucleus and DNA fragmentation, and production of MVC11 declined markedly after prolonged culture. On the other hand, M1 did not induce prominent apoptosis and maintained high virus titers. In primary mouse pulmonary epithelial cell cultures, M1 replicated rather slowly to reach maximum level of virus production at 3 days postinfection, and high levels of virus production were maintained thereafter without causing apoptosis. In contrast, MVC11, which produced 20 times more progeny virus than M1 at 1 day postinfection, induced a high degree of apoptotic cell death before the virus replication cycle was completed. Accordingly, the production of progeny virus was strongly inhibited thereafter. In the lungs of mice infected with MVC11, virus antigens and signals of DNA fragmentation detected by the in situ terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling technique colocalized in bronchial epithelial cells, clearly demonstrating that infection by MVC11 triggered apoptosis in vivo as well as in vitro. These results suggest the possibility that induction of apoptosis by MVC11 plays an important role in attenuation of mouse pathogenicity by restricting progeny virus production in the lung. The C protein was shown to have the capacity to induce apoptosis, and the increased level of the C protein in MVC11-infected cells was considered to account partly, if not entirely, for the induction of apoptosis.


Asunto(s)
Apoptosis , Mutación , Infecciones por Respirovirus/virología , Respirovirus/patogenicidad , Animales , Línea Celular , Células Cultivadas , Células Epiteliales/virología , Pulmón/patología , Pulmón/virología , Macaca mulatta , Ratones , Respirovirus/genética , Respirovirus/crecimiento & desarrollo , Infecciones por Respirovirus/patología , Proteínas Virales/fisiología
20.
Virology ; 238(2): 424-31, 1997 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9400614

RESUMEN

A mutant Sendai virus (SevMVC), which grows much better than its progenitor virus (SeVM) in cell culture, but, in strong contrast to SeVM, is totally avirulent for mice, has been described. SeVMVC contains two amino acid substitutions relative to SeVM, namely, F170S in the C protein and E2050A in the L protein. We have examined which substitutions were responsible for the above phenotypes by exchanging the C gene of our reference strain Z with those of SeVH (another reference strain), SeVM, and SeVMVC, in turn. We have found that the F170S mutation in the CMVC protein is responsible both for enhanced replication in cell culture and for avirulence in mice. Avirulence appeared to be due to restricted viral replication primarily after day 1, implicating some aspect of innate immunity in this process. The SeV C proteins thus appear to be required for multiple cycles of replication in mice.


Asunto(s)
Respirovirus/patogenicidad , Proteínas Virales/fisiología , Animales , Células Cultivadas , Ratones , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mutación Puntual , Respirovirus/genética , Respirovirus/crecimiento & desarrollo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA